59
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      A Diagnostics Platform for the Integrated Mapping, Monitoring, and Surveillance of Neglected Tropical Diseases: Rationale and Target Product Profiles

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background Control and elimination strategies for trachoma, lymphatic filariasis, onchocerciasis, schistosomiasis, ascariasis, trichuriasis and hookworm infection have striking similarities, including the use of periodic mass drug administration (MDA). Because these diseases tend to be co-endemic in the poorest communities of the poorest countries, such that multiple NTDs are frequently found not just in the same populations but within the same individuals [1], it has been suggested that mapping, treatment, impact monitoring, and post-elimination surveillance could be coordinated to better utilise limited human and financial resources. Although many programmes now distribute multiple anthelmintics simultaneously, progress in integrating mapping [2], [3], [4], monitoring, and surveillance [5] activities has been slow [6]. Ideally, population sampling strategies, fieldwork protocols, and sample types (e.g., blood or urine) could all be harmonised between diseases to increase population compliance, simplify overall survey procedures, and decrease costs. For each of these diseases, current diagnostic tools are imperfect (Table S1A), especially for areas with low prevalence. A cost-effective strategy for improved tool development would incorporate integration of diagnostic strategies from the outset [7], [8]. To review available methods for population-based assessment of NTDs, develop target product profiles for tools to monitor infection burden, and consider how those tools would be used in the context of disease elimination programmes, the London School of Hygiene & Tropical Medicine (LSHTM), in collaboration with the World Health Organization, held a consultation at LSHTM from July 19–20, 2010. Participants included disease experts, laboratory and field scientists, authorities on diagnostics, control programme managers, mathematical modellers, and health economists. By bringing together, for the first time, individuals with such a broad spectrum of intersecting disease- and discipline-specific interests to consider issues surrounding integration of diagnostic systems, the consultation aimed to improve on the usual vertical approach to tropical diseases research, encouraging formulation of an innovative approach. This article summarises that consultation's outcomes, suggests target product profiles and a list of immediate research priorities, and drafts a road map for future efforts. We argue for development of a multiplex platform for NTD mapping, monitoring, and surveillance, and suggest changes to policy that might ensue if such a system were to become available. Evolution of Diagnostic Needs with Successful Programme Implementation We conceptualise four time points or periods at which disease elimination programmes require diagnostics: Mapping to establish baseline disease prevalence, facilitating targeting of interventions. Impact monitoring after interventions have commenced. The stopping decision, which determines whether the pre-defined elimination target has been reached, allowing discontinuation of interventions. Post-elimination surveillance after intervention has ceased. Mapping and impact monitoring may require both qualitative and quantitative data from each individual sampled, in order to generate information about the prevalence and intensity of infection. As the prevalence falls with successful control interventions, the intensity of infection also falls. Therefore, to detect the last remaining infections, a more sensitive test may be required. However, for some NTDs (e.g., trachoma), individuals with a low pathogen load are unlikely to transmit infection, so sensitivity is less important. Specificity becomes more important as disease prevalence decreases and is an absolute requirement in certifying elimination [5]. To enable meaningful interpretation of the effect of interventions, the sensitivity and specificity of diagnostic tools used in mapping should be about the same as the sensitivity and specificity of tools used in impact monitoring. One rational difference between mapping and impact monitoring may be the commissioning (in environments where high baseline infection prevalence is expected) of mapping surveys enrolling the same number of clusters over a larger area, in order to save resources. For example, province- or region-level baseline assessments of trachoma prevalence are now accepted for the purposes of requesting donated azithromycin in areas where active trachoma prevalence in 1–9-year-olds is expected (and then proven) to be higher than 10%; subsequent impact monitoring is performed at district level. Ideally, stopping decisions would be based on documentation of the absence of transmission. In practice, however, these decisions are often made once there is documentation of absence of current or previous infection in a sentinel population, such as children born after the time transmission is believed to have been interrupted [9]. Stopping decisions require data generated using diagnostic tools whose specificity is at least as high as those used for mapping, to avoid unnecessarily prolonging MDA. Tool sensitivity is also crucial here to avoid premature MDA cessation and later rebound in infection prevalence. An assay considered adequate for mapping when prevalence is high may have inadequate sensitivity to detect infection in areas with low infection intensity. Stopping decisions are made during the process of impact monitoring, and also mark the commencement of post-elimination surveillance. Data generated to inform stopping decisions should therefore provide useful comparators against both impact monitoring data and data that will subsequently be collected as part of surveillance activities. In general, programmes are likely to require antigen- or nucleic acid–detection assays (to determine prevalence of current infection) for mapping and impact monitoring prior to elimination, and a combination of assays detecting antigens (or nucleic acid) and antibodies (to assess prevalence of exposure in particular population subsets) for post-elimination surveillance. For the worm infections, reliance on detection of transmission stages (eggs, microfilariae) becomes more problematic as the elimination endpoint is approached, since (other than for ascariasis) this will only identify hosts infected with both male and female adults. Specific detection of IgG subtypes may be useful in some cases, particularly if applied at population level: for example, IgG4 responses are characteristic of chronic helminth infections, and titres decline following successful therapy in lymphatic filariasis, onchocerciasis, schistosomiasis, and strongyloidiasis. Monitoring vector, intermediate host, or non-human reservoir [10] populations for the presence of parasites may be important in confirming elimination of infection. Apart from performance characteristics, it is important to consider the operational characteristics of an assay. Large population-based surveys may require tests that can be batched for high throughput. Point of care tests, which generally detect antigen or antibody in dipstick or card format, are relatively cheap, require little formal operator training, and can be performed in the community [11]. They are of particular use when programme personnel need to make immediate decisions as to whether intervention is required. This is helpful when individual patients are being assessed. However, for MDA, where decisions are needed on whether or not to treat whole communities or districts, laboratory-based tests are probably adequate, provided samples are easy to collect (e.g., fingerprick) and transport (e.g., dried blood spots). Target Product Profiles and Immediate Research Priorities Target product profiles for lymphatic filariasis, trachoma, schistosomiasis, onchocerciasis, and soil-transmitted helminths are shown for the mapping and impact monitoring phases in Table 1, and for the post-elimination surveillance phase (first four diseases only) in Table 2. The tables consider only the needs for diagnostic tools in mapping, monitoring, and surveillance of human infection because we see these as priorities for any first-generation integrated platform for NTD diagnostics; we have, for the moment, put aside programme requirements for monitoring MDA coverage; measures of morbidity; possible emergence of drug resistance; prevalence of infection in vectors, intermediate hosts, or reservoir animals; and force of transmission through environmental sampling. 10.1371/journal.pntd.0001746.t001 Table 1 Proposed target product profiles for diagnostic tools for selected NTDs, mapping, and impact monitoring. Characteristic Lymphatic Filariasis Trachoma Schistosomiasis Onchocerciasis Soil-Transmitted Helminths Intended use Mapping, monitoring, and stopping decision Mapping, monitoring, and stopping decision Mapping, monitoring, and stopping decision Mapping, monitoring, and stopping decision Mapping and monitoring Possible target populationa 6–15-year-old children 1–9-year-old children (could be adjusted) 6–15-year-old children plus occupational groups 6–15-year-old children 6–15-year-old children Possible sample types Blood spot Eye swab (other: mouth or nose swab, tears) Blood spot or urine (avoid stool if possible) Blood spot Blood spot or urine (avoid stool if possible) Ideal diagnostic marker Parasite antigen C. trachomatis antigen Species-specific antigen OR pan-genus antigen Parasite antigen Parasite antigen Ideal test format POC or high throughput laboratory assay POC or high throughput laboratory assay POC assay POC or high throughput laboratory assay POC assay Availability of ideal diagnostic marker Available but not right format, low reliability, high cost, and temperature sensitive Available but not right format Not yet available Not yet available. IgG4 antibody may be a reasonable proxy Not yet available Required performance characteristics 95% sensitive; W. bancrofti-specific >50% sensitive, 99.5% specific >50% sensitive, 99.5% specific >50% sensitive, 99.5% specific >50% sensitive, 99.5% specific Comparator assay (current reference standard) Night blood micro-filaraemia Quantitative PCR Kato-Katz (multiple slides and multiple days) and/or urine filtration Skin snips to detect micro-filariae Kato-Katz (multiple slides and multiple days) Possible sampling strategies PBPS/LQAS, school based, sentinel sites PBPS/LQAS, home based, sentinel sites PBPS/LQAS, school based, 50/school, increasing with control PBPS/LQAS PBPS/LQAS, school based LQAS, lot quality assurance sampling; NTDs, neglected tropical diseases; PBPS, population-based prevalence survey; PCR, polymerase chain reaction; POC, point of care. a Based on peak infection prevalence, convenience, or both. 10.1371/journal.pntd.0001746.t002 Table 2 Proposed target product profiles for diagnostic tools for selected NTDs, post-elimination surveillance.a Characteristic Lymphatic Filariasis Trachoma Schistosomiasis Onchocerciasis Intended use Post-elimination incidence of infection Post-elimination incidence of infection Post-elimination incidence of infection Post-elimination incidence of infection Possible target population Children born after transmission interruption Children born after transmission interruption Children born after transmission interruption Children born after transmission interruption Possible sample types Blood spot Blood spot Blood spot or urine (avoid stool if possible) Blood spot Ideal diagnostic marker Antibody Antibody to a conserved species-specific epitope of MOMP Antibody Ov16 antibody Availability of ideal diagnostic marker Not available Libraries available In development Available, but additional validation needed Ideal test format High throughput laboratory assay High throughput laboratory assay High throughput laboratory assay High throughput laboratory assay Population infection thresholds (for stopping MDA) 1% Not defined 10% of school-aged children 1/3,000 Probable sampling strategy PBPS PBPS PBPS or school surveys (or sentinel occupations) PBPS a Schistosomiasis is included in this table because several countries have programmes to eliminate this disease [18], [19]. The soil-transmitted helminth infections are not included because (as for schistosomiasis in most endemic states) the current goal is prevention of morbidity in school-aged children through periodic high-coverage MDA. ICT, immunochromatographic card test; LF, lymphatic filariasis; MDA, mass drug administration; MOMP, major outer membrane protein of C. trachomatis; NTDs, neglected tropical diseases; PBPS, population-based prevalence survey. The target product profiles that we set out here are aspirational. Some tests (e.g., antigen assay for W. bancrofti [Table 1] or Ov16 antibody assay in previously onchocerciasis-endemic areas [Table 2]) appear close to being validated for programme use, while for others, numerous technical hurdles remain. For this reason, we expect some of our target product profiles—particularly blood- or urine-based antigen detection tests for the soil-transmitted helminthiases—to be controversial. However, there is presently at least one commercially available ELISA kit to detect IgG directed against Ascaris lumbricoides in human serum: it should be possible to develop a test to detect the antigen driving that antibody response. If such antigens only circulate briefly in the early part of the Ascaris life cycle, this may actually be helpful in interpreting test results at community level, since antigen detection will indicate the presence of ongoing transmission. Immediate research priorities are shown in Table 3. 10.1371/journal.pntd.0001746.t003 Table 3 Immediate research priorities. Disease Research Goal Feasibility (0–10a: 0, Impossible; 10, Inevitable) Impact if Achieved (0–10a: 0, None; 10, Massive) Lymphatic filariasis Development of antigen tests to usable/reliable format 9 8 if ≤USD 0.50 Development and validation of tests (e.g., IgG4-subclass antibody detection tests using recombinant Bm14, BmR1, WbSXP, and W. bancrofti-specific antigens [20] or PCR-based detection of parasite DNA in homogenised mosquitoes [21]) useful for post-elimination surveillance, with accompanying standardised survey methodologies 9 8 if ≤USD 0.50 Trachoma Development of a test for ocular C. trachomatis infection [22] able to maintain specificity at high temperatures and low humidity [23] 9 8 if ≤USD 0.50 Development of eye/nose swab-, saliva-, or blood-based anti-C. trachomatis antibody test and exploration of the impact of successful trachoma control on antibody profiles in endemic populations 3 5 Development and validation of a school-based survey protocol (need threshold minimum school attendance) 7 8 Schistosomiasis Development of antigen [24] or antibody [25] isotype combination(s) useful in high and low transmission intensity environments, able to distinguish current from past infection 8 9 Development of antigen or antibody isotype combination(s) to distinguish between different species 8 4 Development of serum markers of morbidity 6 8 Soil-transmitted helminthiases Development of reliable blood- or urine-based assays for detection of current infection 4 9 Development of serum markers of morbidity 6 8 Onchocerciasis Development of a quantitative antigen test for use in endemic areas in Africa and validation of Ov16 antibody test for demonstrating interruption of transmission in Africa 5 8 Development of a test for loaiasis 5 9 a Determined by expert consensus. Discussion Trachoma, lymphatic filariasis, schistosomiasis, onchocerciasis, and soil-transmitted helminth infections are found in overlapping populations; are controlled through broadly similar, often complementary, strategies involving MDA; and are mapped and monitored by sampling individuals from the population-at-risk using strategies that are also broadly similar but different in detail. Programmes for their control and elimination require improved diagnostic tools to guide decisions on the required intensity, frequency, and duration of intervention and to conduct surveillance for re-emergence of infection after elimination. Similarities between target product profiles (Tables 1 and 2) suggest the feasibility and desirability of integration of diagnostic approaches. In many areas in which NTDs are highly endemic, basic health infrastructure is sparse or non-existent, and there are few trained personnel. Local laboratories may not have access to refrigeration, reliable power, or piped water; have highly variable capacity for performing diagnostic assays; and the capacity they do have is in general insufficient to meet existing diagnostic requirements of local clinical services. They are therefore ill-equipped to take on the extra burden of generating data to feed into NTD elimination programmes without provision of additional money, staff, training, equipment, reagents, and utilities—or robust technologies that could perform well despite limitations to supply of these resources. The ideal integrated system might therefore be a portable, self-contained diagnostics platform, capable of performing multiplex assays for several infections of interest on one or a small number of sample types. A system employing microfluidics (“lab-on-a-chip”) [12], [13], [14] technology could fulfil these requirements. The platform should be able to simultaneously undertake multiple roles in different NTD control programmes, each of which might be at various points of evolution within a given population. For example, in a district that had been hyperendemic at baseline for trachoma, soil-transmitted helminths, and lymphatic filariasis but in which interventions had already been in progress for a number of years, the platform would be capable of accurately detecting reductions in ocular C. trachomatis infection, whilst simultaneously measuring the prevalence of soil-transmitted helminth infection and monitoring for post-elimination re-emergence of lymphatic filariasis. Since diseases of potential interest will vary from one population to the next, a modular format would provide opportunities to swap diagnostic capacity for particular infections in and out of the platform according to global, regional, or local priority. For example, in onchocerciasis-endemic areas, the capacity to test for loaiasis at the same time as measuring the prevalence of O. volvulus infection would benefit programmes [15]. Equally, the platform should be adaptable for the assessment of the community prevalence of HIV infection, malaria parasitaemia or anti-malaria antibody, and/or seroprevalence of antibodies to measles, rubella, or hepatitis B surface antigen following vaccination campaigns. Our vision can be conceptualised as the delivery of two linked components: a hardware module, on which samples will be processed, and various elements of software, including both the assays themselves and the algorithms to guide their use in the field. To ensure that any new technologies are ready for both registration and end use, field personnel, programme managers, regulatory agencies, ministries of health, and other key stakeholders should be involved in platform development and evaluation. In addition to the potential savings to existing vertical control programmes that would become possible through integration of diagnostic tools, this approach has several other potential advantages. First, it makes conducting surveys to rule out specific diseases easier and more cost-effective. This can occasionally yield surprising results. In Burundi in 2007, examination for trachoma was included alongside fieldwork conducted nationally to estimate the prevalences of schistosomiasis and soil-transmitted helminths, in order to confirm the long-held belief that Burundi was trachoma-free. Active trachoma was found in children throughout the country, and trachoma control activities including azithromycin MDA commenced in 2011 in three districts. Second, proof-of-concept of an integrated diagnostics platform could facilitate programme planning for other infections for which control strategies are in the early stages of development. An October 2009 WHO expert consultation discussed recent work piloting taeniasis elimination in Peru and possible MDA approaches for food-borne trematode infections. These diseases may have global control initiatives developed in the foreseeable future. Third, establishing capacity for reliable diagnosis of what have hitherto been the most neglected diseases could catalyse a frame-shift in the global health community's vision of developing world laboratory science. A diagnostics platform that could be configured to generate community- or individual-level data for any of the infections already mentioned as well as perform tests for (for example) sexually transmitted infections, human African trypanosomiasis, or leishmaniasis would represent a game-changing advance in the fight against infectious diseases. World Health Assembly Resolution 60.29 on Health Technologies recognizes that medical devices are indispensable tools for prevention, diagnosis, treatment, and rehabilitation in health care [16]. It is widely accepted that the availability of, and access to, appropriate and affordable health technologies in low- and middle-income countries remain inadequate. In 2010, WHO held the first Global Forum on Medical Devices [17], which featured selected technological innovations that could improve global health. The innovators identified financing, manufacturing partners, and distribution channels as their top three challenges in getting their technologies into resource-limited settings. WHO undertook to continue to interact with industry, funding agencies, academia, and international organizations to raise awareness of the need to design, produce, and commercialize innovative, accessible, and robust technologies which address the needs of health systems particularly in low-resource settings. The development, evaluation, and deployment of an integrated platform to monitor progress towards NTD elimination would be consistent with this WHO vision. Supporting Information Table S1 Performance against the ASSURED criteria [11] of existing diagnostic tools for the neglected tropical diseases employing mass drug administration. (DOC) Click here for additional data file.

          Related collections

          Most cited references16

          • Record: found
          • Abstract: found
          • Article: not found

          A strategy to control transmission of Schistosoma japonicum in China.

          Schistosoma japonicum causes an infection involving humans, livestock, and snails and is a significant cause of morbidity in China. We evaluated a comprehensive control strategy in two intervention villages and two control villages along Poyang Lake in the southeastern province of Jiangxi, where annual synchronous chemotherapy is routinely used. New interventions, implemented from 2005 through 2007, included removing cattle from snail-infested grasslands, providing farmers with mechanized farm equipment, improving sanitation by supplying tap water and building lavatories and latrines, providing boats with fecal-matter containers, and implementing an intensive health-education program. During the intervention period, we observed changes in S. japonicum infection in humans, measured the rate of infection in snails, and tested the infectivity of lake water in mice. After three transmission seasons, the rate of infection in humans decreased to less than 1.0% in the intervention villages, from 11.3% to 0.7% in one village and from 4.0% to 0.9% in the other (P<0.001 for both comparisons). The rate of infection in humans in control villages fluctuated but remained at baseline levels. In intervention villages, the percentage of sampling sites with infected snails decreased from 2.2% to 0.1% in one grassland area and from 0.3% to no infection in the other (P<0.001 for both comparisons). The rate of infection in mice after exposure to lake water decreased from 79% to no infection (P<0.001). A comprehensive control strategy based on interventions to reduce the rate of transmission of S. japonicum infection from cattle and humans to snails was highly effective. These interventions have been adopted as the national strategy to control schistosomiasis in China. 2009 Massachusetts Medical Society
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Nano/Microfluidics for diagnosis of infectious diseases in developing countries.

            Nano/Microfluidic technologies are emerging as powerful enabling tools for diagnosis and monitoring of infectious diseases in both developed and developing countries. Miniaturized nano/microfluidic platforms that precisely manipulate small fluid volumes can be used to enable medical diagnosis in a more rapid and accurate manner. In particular, these nano/microfluidic diagnostic technologies are potentially applicable to global health applications, since they are disposable, inexpensive, portable, and easy-to-use for detection of infectious diseases. In this paper, we review recent advances in nano/microfluidic technologies for clinical point-of-care applications at resource-limited settings in developing countries. Copyright 2009 Elsevier B.V. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Microfluidics without pumps: reinventing the T-sensor and H-filter in paper networks.

              Conventional microfluidic devices typically require highly precise pumps or pneumatic control systems, which add considerable cost and the requirement for power. These restrictions have limited the adoption of microfluidic technologies for point-of-care applications. Paper networks provide an extremely low-cost and pumpless alternative to conventional microfluidic devices by generating fluid transport through capillarity. We revisit well-known microfluidic devices for hydrodynamic focusing, sized-based extraction of molecules from complex mixtures, micromixing, and dilution, and demonstrate that paper-based devices can replace their expensive conventional microfluidic counterparts.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Negl Trop Dis
                PLoS Negl Trop Dis
                plos
                plosntds
                PLoS Neglected Tropical Diseases
                Public Library of Science (San Francisco, USA )
                1935-2727
                1935-2735
                July 2012
                31 July 2012
                : 6
                : 7
                : e1746
                Affiliations
                [1 ]London School of Hygiene & Tropical Medicine, London, United Kingdom
                [2 ]World Health Organization, Geneva, Switzerland
                [3 ]Imperial College London, London, United Kingdom
                [4 ]National Institute for Parasitic Diseases, Shanghai, People's Republic of China
                [5 ]Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
                [6 ]Institut National D'Hygiène, Rabat, Morocco
                [7 ]Liverpool School of Tropical Medicine, Liverpool, United Kingdom
                Queensland Institute for Medical Research, Australia
                Author notes

                The authors have declared that no competing interests exist.

                Article
                PNTD-D-11-00459
                10.1371/journal.pntd.0001746
                3409112
                22860146
                3e260262-2e4f-48cb-8842-c5243caf9ace
                Copyright @ 2012

                This is an open-access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.

                History
                Page count
                Pages: 5
                Funding
                The consultation that laid the foundation for this manuscript was supported by the World Health Organization. The funder had no role in the decision to publish, or in preparation of the manuscript, other than the individual contributions recognised by authorship of the paper.
                Categories
                Policy Platform
                Medicine
                Global Health
                Infectious Diseases
                Neglected Tropical Diseases
                Ascariasis
                Hookworm
                Lymphatic Filariasis
                Onchocerciasis
                Schistosomiasis
                Soil-Transmitted Helminths
                Trachoma
                Trichuriasis

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article